GPX3 Human

Glutathione Peroxidase 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Expression and Localization

GPX3 is primarily synthesized in kidney proximal tubular cells and secreted into plasma . Its expression is also detected in liver, heart, lung, and thyroid tissues . Unique features include:

TissueExpression LevelKey Function
KidneyHighestPlasma GPX3 synthesis; binds basement membranes of cortical tubules
LiverModerateOxidative stress regulation in extracellular fluids
LungLowProtection against ROS in alveolar spaces

In vivo studies using Gpx3 knockout mice revealed that kidney basement membranes selectively bind GPX3, suggesting localized antioxidant activity in renal peritubular spaces .

Cancer-Related Roles

GPX3 exhibits dual roles in tumorigenesis:

Cancer TypeExpression TrendPrognostic ImpactImmune Correlation
GastricDownregulatedUnfavorable prognosis Positive correlation with CD8+ T cells
BreastUpregulatedPromotes metastasis under oxidative stress Modulates macrophage infiltration

In gastric cancer, low GPX3 expression is associated with advanced tumor stages (T3/T4) and immune evasion . Conversely, in advanced breast cancer, GPX3 enhances metastasis by scavenging ROS during anoikis .

ROS Modulation

GPX3 regulates ROS levels in mitochondria and plasma, influencing signaling pathways such as:

  • NF-κB: Controls inflammation and apoptosis .

  • Wnt/β-catenin: Regulates cell proliferation and differentiation .

Epigenetic and Post-Transcriptional Regulation

  • Promoter Methylation: Silencing in cancers (e.g., thyroid, hepatocellular) .

  • miRNA/lncRNA Interactions: DUBR/hsa-miR-502-3p/GPX3 axis modulates expression .

In Vitro Studies

Cell LineGPX3 ManipulationOutcomeSource
Ovarian (Ovcar-4)KnockdownReduced proliferation, enhanced apoptosis
Breast (MDA-MB-231)KnockdownInhibited migration under oxidative stress

In Vivo Findings

  • Gpx3 knockout mice showed no overt phenotype, suggesting compensation by other selenoproteins .

  • Plasma GPX3 levels drop post-nephrectomy and recover after kidney transplantation .

Production Challenges

Recombinant GPX3 synthesis in E. coli is hindered by:

  • Selenocysteine Incorporation: Requires auxotrophic strains and SECIS elements .

  • Disulfide Bridges: Absence of Cys residues complicates folding .

Therapeutic Potential

GPX3’s role in cancer progression highlights its potential as a biomarker or therapeutic target. Further studies are needed to:

  • Elucidate tissue-specific regulatory mechanisms.

  • Explore GPX3-based strategies to modulate tumor microenvironments.

Product Specs

Introduction
Glutathione peroxidase 3 (GPX3), a member of the glutathione peroxidase family, plays a crucial role in detoxifying hydrogen peroxide. It protects cells and enzymes from oxidative damage by catalyzing the reduction of hydrogen peroxide, lipid peroxides, and organic hydroperoxide using glutathione. GPX3 is notable for containing selenocysteine at its active site, a rare occurrence in higher vertebrates. This selenocysteine is encoded by the stop codon TGA.
Description
Recombinant human GPX3, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 227 amino acids (21-226), resulting in a molecular weight of 25.7 kDa. The protein includes a 21 amino acid His-tag at the N-terminus and undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The GPX3 solution is supplied in a buffer containing 20mM Tris-HCl (pH 7.5), 40% glycerol, 0.15M NaCl, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), the product can be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the product is greater than 85% as determined by SDS-PAGE analysis.
Synonyms
Glutathione peroxidase 3, GPx-3, GSHPx-3, Extracellular glutathione peroxidase, Plasma glutathione peroxidase, GPx-P, GSHPx-P, GPX3, GPXP.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MQSRGQEKSK MDCHGGISGT IYEYGALTID GEEYIPFKQY AGKYVLFVNV ASYCGLTGQY IELNALQEEL APFGLVILGF PCNQFGKQEP GENSEILPTL KYVRPGGGFV PNFQLFEKGD VNGEKEQKFY TFLKNSCPPT SELLGTSDRL FWEPMKVHDI RWNFEKFLVG PDGIPIMRWH HRTTVSNVKM DILSYMRRQA ALGVKRK.

Q&A

What is GPX3 and what is its primary function in human biology?

GPX3, or glutathione peroxidase 3, is a unique glycosylated extracellular isoform of the glutathione peroxidase family that catalyzes the reduction of various hydroperoxides to their corresponding alcohols using GSH as a substrate . While predominantly expressed in the kidney, GPX3 is also found in multiple other tissues including adipose tissue, where it plays significant roles in metabolism . The enzyme contains selenocysteine (Sec) in its active center, which is incorporated through a specific biochemical mechanism crucial for its catalytic function . Unlike other GPx family members, GPX3 exists as a tetramer in its native state, and this quaternary structure appears essential for optimal enzymatic activity . Its primary function involves neutralizing reactive oxygen species (ROS), thereby providing protection against oxidative stress-induced cellular damage across various tissues and physiological systems .

What are the structural characteristics of human GPX3?

Human GPX3 possesses several distinct structural characteristics that contribute to its unique enzymatic properties. The protein exists as a tetramer in its native state, which is critical for its optimal catalytic function . The active site contains selenocysteine (Sec), a specialized amino acid that functions as the primary catalytic residue for peroxide reduction . GPX3 is the only known glycosylated member of the GPx family, with post-translational glycosylation appearing to influence its catalytic efficiency and stability . Studies using recombinant human GPX3 mutants have revealed that alterations to the cysteine residues affect quaternary structure formation, typically resulting in monomeric forms with reduced catalytic capability compared to the native tetrameric enzyme . The human GPX3 gene spans approximately 10 kb on chromosome 5 at region q32 and is organized into five exons, with the first exon encoding the signal peptide necessary for extracellular secretion . These structural features collectively contribute to GPX3's specialized function as an extracellular peroxidase with unique substrate preferences and activity profiles.

How does GPX3 regulate insulin sensitivity in adipose tissue?

GPX3 has emerged as a critical regulator of insulin sensitivity in adipose tissue through multiple interrelated mechanisms. Research has demonstrated that GPX3 expression positively correlates with insulin receptor (IR) expression in adipose tissue, suggesting a direct regulatory relationship between these proteins . In animal models of insulin resistance, including ob/ob mice and high-fat diet models, GPX3 expression is significantly reduced in white adipose tissue, particularly gonadal adipose deposits, mirroring changes in insulin receptor levels . The mechanism appears to involve GPX3-mediated activation of the transcription factor Sp1, which enhances insulin receptor expression in adipocytes . Interestingly, selenium supplementation increases GPX3 expression in adipose tissue, subsequently improving insulin sensitivity, enhancing adipocyte differentiation, and reducing tissue inflammation . This selenium-GPX3-insulin receptor axis represents a potentially novel therapeutic target for metabolic disorders. Human studies have further validated these findings, showing decreased GPX3 expression in adipose tissue from obese and insulin-resistant patients compared to normal-weight controls, suggesting the translational relevance of these mechanistic insights .

What role does GPX3 play in reproductive biology and spermatogenesis?

GPX3 plays a crucial regulatory role in human spermatogenesis through modulation of spermatogonial stem cell (SSC) fate determination. Recent research has demonstrated GPX3 expression in human SSCs, where it functions as a key regulator of proliferation, DNA synthesis, and apoptosis control . Experimental GPX3 knockdown in human SSC lines leads to decreased proliferation, reduced DNA synthesis, and diminished cyclin B1 levels, collectively impairing replicative capacity . Moreover, GPX3 silencing enhances early apoptosis in these cells, suggesting dual roles in both promoting cell cycle progression and suppressing cell death . Mechanistically, GPX3 appears to function through interaction with CXCL10, as demonstrated by co-immunoprecipitation studies and functional analyses showing that CXCL10 knockdown phenocopies GPX3 silencing effects . This GPX3-CXCL10 regulatory axis represents a potentially novel pathway in human spermatogenesis. The identification of this pathway provides new insights into the molecular mechanisms governing male fertility and offers potential therapeutic targets for addressing certain forms of male infertility associated with spermatogenic dysfunction or oxidative stress-induced reproductive impairment.

What evidence links GPX3 to neurodegenerative disorders?

Emerging evidence strongly implicates GPX3 in the pathogenesis of several neurodegenerative disorders, particularly amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD). Cross-ethnic genetic analyses have identified significant association between the GPX3-TNIP1 locus and ALS risk, with rs10463311 spanning this region achieving genome-wide significance (p = 1.3 × 10−8) and replication support from independent Australian cohorts . Mechanistically, GPX3 interacts with SOD1, a well-established ALS-associated gene, suggesting functional integration within critical oxidative stress response pathways relevant to motor neuron survival . In parallel, recent research has identified GPX3 as a promising therapeutic target in the shared molecular mechanisms between traumatic brain injury (TBI) and Parkinson's disease . Differential gene expression analyses across multiple datasets have positioned GPX3 as a hub gene within networks connecting TBI pathology to subsequent neurodegenerative risk . These findings are particularly significant given the epidemiological association between moderate-to-severe TBI and elevated PD risk, providing a potential mechanistic basis for this relationship . The involvement of GPX3 in multiple neurodegenerative contexts suggests its fundamental role in neuronal redox homeostasis and neuroprotection, positioning it as both a biomarker and potential therapeutic target for these conditions.

How is GPX3 dysregulation implicated in cancer pathogenesis?

GPX3 dysregulation has been increasingly implicated in cancer pathogenesis, with particular evidence in prostate cancer models. Studies have documented reduced GPX3 expression in human prostate cancer tissues, suggesting potential tumor suppressor functions . In experimental models, loss of GPX3 in Nkx3.1 knockout mice (a model for prostatic intraepithelial neoplasia) influences reactive oxygen species (ROS) levels and contributes to prostatic hyperplasia progression . Specifically, double knockout mice (Nkx3.1−/−; Gpx3−/−) exhibit altered expression patterns of oxidative stress-associated genes including SOD3, iNOS, Hmox, and CISD2, with initial compensatory upregulation at 4 months followed by decline at later timepoints, suggesting progressive oxidative stress defense failure . This temporal pattern aligns with the concept that cancer cells often experience initial adaptive responses to oxidative stress that eventually become overwhelmed as disease progresses. The mechanistic relationship between GPX3 deficiency and malignant transformation appears to involve chronic ROS accumulation, which can damage DNA, modify signaling pathways, and promote genomic instability. These findings collectively position GPX3 as a potential tumor suppressor in prostatic and potentially other tissues, where its loss may contribute to oxidative stress-driven carcinogenesis through disruption of redox homeostasis.

What are the optimal approaches for measuring GPX3 expression and activity?

Accurate measurement of GPX3 expression and activity requires careful consideration of methodological approaches tailored to the specific research question. For quantifying GPX3 gene expression, quantitative real-time PCR (qRT-PCR) offers high sensitivity using validated primers targeting conserved regions of GPX3 mRNA, with normalization to appropriate housekeeping genes essential for reliable comparisons across different physiological states . At the protein level, western blotting with specific anti-GPX3 antibodies enables semi-quantitative assessment of total protein levels, while immunohistochemistry and immunofluorescence provide valuable spatial information about tissue and cellular distribution patterns . Enzyme activity assays typically employ spectrophotometric methods measuring NADPH oxidation coupled to glutathione reductase, though these require careful optimization to distinguish GPX3 activity from other glutathione peroxidases . For investigating GPX3's selenoprotein nature, incorporation of radiolabeled selenium (75Se) can provide insights into selenoprotein synthesis rates and efficiency . In cell culture systems, selenite supplementation at approximately 200 nM has been demonstrated as optimal for GPX3 regulation, representing an important consideration for in vitro studies . Recent methodological advances include mass spectrometry-based approaches for precise quantification of GPX3 protein levels and potential post-translational modifications that may influence enzymatic function in different physiological and pathological contexts.

What experimental models are most appropriate for studying GPX3 function?

Selection of appropriate experimental models is crucial for investigating GPX3 function across different physiological contexts. Cell culture systems, particularly the 3T3-L1 preadipocyte line, have proven valuable for studying GPX3's role in adipocyte differentiation and insulin signaling, with selenite supplementation (optimally 200 nM) enabling manipulation of GPX3 expression levels . For reproductive biology research, established human spermatogonial stem cell lines that maintain phenotypic features of primary SSCs provide suitable models for investigating GPX3's role in spermatogenesis . Genetic manipulation approaches, including siRNA knockdown and shRNA stable transfection, allow for targeted GPX3 silencing to assess functional consequences in these cellular systems . In vivo, several mouse models have proven informative, including selenium-enriched high-fat diet models for metabolic studies and Gpx3 knockout mice (Gpx3−/−) for investigating systemic effects of GPX3 deficiency . The combined Nkx3.1−/−; Gpx3−/− double knockout mouse represents a specialized model for studying GPX3's role in prostatic hyperplasia and cancer progression . For neurodegenerative disease research, cross-ethnic genome-wide association studies have identified significant GPX3 variants, which can be further characterized using transgenic models expressing these variants . When selecting experimental models, researchers should consider tissue-specific expression patterns, potential compensatory mechanisms from other glutathione peroxidases, and species differences in selenoprotein biology that may affect translational relevance.

How can researchers effectively manipulate GPX3 levels in experimental systems?

Researchers have several sophisticated approaches available for manipulating GPX3 levels in experimental systems, each with specific advantages and limitations. RNA interference techniques, including siRNA transfection for transient knockdown and shRNA for stable suppression, have been successfully employed to reduce GPX3 expression in various cell types including 3T3-L1 preadipocytes and human spermatogonial stem cell lines . For enhancing GPX3 expression, selenium supplementation represents a physiologically relevant approach, with 200 nM selenite identified as optimal for GPX3 regulation in cell culture systems . More specific manipulation can be achieved through plasmid-based overexpression of wild-type or mutant GPX3 variants, which allows for structure-function relationship studies . The CRISPR-Cas9 gene editing system offers precise genomic modification for creating cellular or animal models with GPX3 knockout, knock-in, or specific mutations. For in vivo studies, Gpx3 knockout mice (Gpx3−/−) provide a valuable model for studying systemic GPX3 deficiency, while conditional knockout approaches using Cre-loxP systems enable tissue-specific GPX3 deletion to address questions about local versus systemic effects . When manipulating GPX3 levels, researchers should monitor potential compensatory changes in other antioxidant systems and consider the broader selenotranscriptome context, as alterations in selenium availability may affect multiple selenoproteins simultaneously, potentially confounding experimental interpretations.

What is the potential of GPX3 as a biomarker or therapeutic target?

GPX3 demonstrates significant potential as both a biomarker and therapeutic target across multiple disease contexts. In metabolic disorders, serum and adipose tissue GPX3 levels correlate with insulin sensitivity, suggesting utility as a biomarker for metabolic dysfunction and potential treatment response . The demonstrated ability of selenium supplementation to enhance GPX3 expression and subsequently improve insulin sensitivity in adipose tissue provides a translational pathway for therapeutic intervention in insulin resistance and obesity . In reproductive medicine, GPX3 expression patterns in seminal plasma might serve as biomarkers for specific forms of male infertility, while targeting the GPX3-CXCL10 axis represents a potential therapeutic approach for enhancing spermatogenesis in certain infertility conditions . For neurodegenerative disorders, the significant association between GPX3-TNIP1 locus variants and ALS risk positions GPX3 as both a potential risk biomarker and therapeutic target . Similarly, GPX3's involvement in shared molecular mechanisms between traumatic brain injury and Parkinson's disease suggests applications in early detection of neurodegeneration risk following TBI and potential neuroprotective interventions . In oncology, reduced GPX3 expression in prostatic hyperplasia and cancer indicates potential diagnostic value, while approaches to restore GPX3 function might offer novel therapeutic strategies for prostate cancer prevention or treatment . The development of recombinant human GPX3 preparations, including unglycosylated variants with retained activity, provides potential biotherapeutic avenues for conditions associated with oxidative stress and GPX3 deficiency .

How do genetic variations in GPX3 influence disease susceptibility?

Genetic variations in GPX3 significantly influence disease susceptibility across multiple pathological contexts through both direct functional alterations and regulatory impacts. In amyotrophic lateral sclerosis (ALS), cross-ethnic meta-analysis has identified a significant association between the rs10463311 variant spanning the GPX3-TNIP1 locus and disease risk (p = 1.3 × 10−8), with replication support from independent cohorts . This finding is mechanistically supported by known interactions between GPX3 and SOD1, a well-established ALS-associated gene . Population-specific differences in linkage disequilibrium patterns enhance gene-mapping resolution in cross-ethnic studies, allowing identification of functional variants that might be missed in single-population analyses . Beyond single nucleotide polymorphisms, epigenetic modifications and copy number variations affecting GPX3 expression contribute to disease risk profiles, particularly in metabolic disorders where adipose tissue GPX3 expression correlates with insulin sensitivity . In reproductive biology, GPX3 variants may influence male fertility through effects on spermatogenesis, as GPX3 regulates proliferation and apoptosis of spermatogonial stem cells . The functional consequences of GPX3 variants may be modulated by environmental factors, particularly selenium availability, creating gene-environment interactions that influence disease susceptibility . Integration of GPX3 genetic data with clinical phenotyping and functional genomics approaches is essential for translating genetic associations into mechanistic understanding and potentially personalized therapeutic strategies.

Product Science Overview

Introduction

Glutathione Peroxidase 3 (GPX3), also known as plasma glutathione peroxidase (GPx-P) or extracellular glutathione peroxidase, is an enzyme encoded by the GPX3 gene in humans . It belongs to the glutathione peroxidase family, which plays a crucial role in protecting cells from oxidative damage by reducing hydrogen peroxide, lipid peroxides, and organic hydroperoxides .

Structure and Function

GPX3 contains a selenocysteine (Sec) residue at its active site, which is essential for its enzymatic activity . The selenocysteine is encoded by the UGA codon, which typically signals translation termination. However, in the case of Sec-containing genes, a specific stem-loop structure in the 3’ untranslated region (UTR), known as the Sec insertion sequence (SECIS), allows the UGA codon to be recognized as a Sec codon instead of a stop signal .

GPX3 functions as an oxidoreductase enzyme, catalyzing the conversion of hydrogen peroxide into water, thereby mitigating oxidative damage . It has a wide thiol specificity, with sources of reducing power including glutathione (GSH), cysteine, mercaptoethanol, and dithiothreitol . In vitro studies have shown that GSH can be completely replaced by reduced homocysteine for GPX3 activity .

Biological Significance

GPX3 is primarily found in the extracellular space, including blood plasma, and is involved in detoxifying hydrogen peroxide in the extracellular environment . It plays a vital role in maintaining the redox balance and protecting cells from oxidative stress . Beyond its antioxidant function, GPX3 is also involved in regulating metabolism, modulating cell growth, inducing apoptosis, and facilitating signal transduction .

Clinical Relevance

Recent studies have highlighted the significance of GPX3 in various non-neoplastic diseases. Aberrant expression of GPX3 has been associated with multiple pathological processes, including cardiovascular diseases, metabolic disorders, and inflammatory conditions . Additionally, GPX3 serves as a significant tumor suppressor in various cancers, making it a potential diagnostic biomarker and therapeutic target .

Recombinant GPX3

Recombinant human GPX3 is produced using recombinant DNA technology, allowing for the large-scale production of this enzyme for research and therapeutic purposes . Recombinant GPX3 retains the same structure and function as the naturally occurring enzyme, making it a valuable tool for studying oxidative stress and developing potential treatments for diseases associated with oxidative damage .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.